Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Nir Skalka is active.

Publication


Featured researches published by Nir Skalka.


Molecular Psychiatry | 2014

The NAP motif of activity-dependent neuroprotective protein (ADNP) regulates dendritic spines through microtubule end binding proteins

Saar Oz; O Kapitansky; Y Ivashco-Pachima; Anna Malishkevich; Eliezer Giladi; Nir Skalka; Rina Rosin-Arbesfeld; L Mittelman; O Segev; J A Hirsch; Illana Gozes

The NAP motif of activity-dependent neuroprotective protein (ADNP) enhanced memory scores in patients suffering from mild cognitive impairment and protected activities of daily living in schizophrenia patients, while fortifying microtubule (MT)-dependent axonal transport, in mice and flies. The question is how does NAP fortify MTs? Our sequence analysis identified the MT end-binding protein (EB1)-interacting motif SxIP (SIP, Ser-Ile-Pro) in ADNP/NAP and showed specific SxIP binding sites in all members of the EB protein family (EB1–3). Others found that EB1 enhancement of neurite outgrowth is attenuated by EB2, while EB3 interacts with postsynaptic density protein 95 (PSD-95) to modulate dendritic plasticity. Here, NAP increased PSD-95 expression in dendritic spines, which was inhibited by EB3 silencing. EB1 or EB3, but not EB2 silencing inhibited NAP-mediated cell protection, which reflected NAP binding specificity. NAPVSKIPQ (SxIP=SKIP), but not NAPVAAAAQ mimicked NAP activity. ADNP, essential for neuronal differentiation and brain formation in mouse, a member of the SWI/SNF chromatin remodeling complex and a major protein mutated in autism and deregulated in schizophrenia in men, showed similar EB interactions, which were enhanced by NAP treatment. The newly identified shared MT target of NAP/ADNP is directly implicated in synaptic plasticity, explaining the breadth and efficiency of neuroprotective/neurotrophic capacities.


Oncogene | 2013

Carboxypeptidase E: a negative regulator of the canonical Wnt signaling pathway

Nir Skalka; Michal Caspi; E Caspi; Y P Loh; Rina Rosin-Arbesfeld

Aberrant activation of the canonical Wnt signal transduction pathway is involved in many diseases including cancer and is especially implicated in the development and progression of colorectal cancer. The key effector protein of the canonical Wnt pathway is β-catenin, which functions with T-cell factor/lymphoid enhancer factor to activate expression of Wnt target genes. In this study, we used a new functional screen based on cell survival in the presence of cDNAs encoding proteins that activate the Wnt pathway thus identifying novel Wnt signaling components. Here we identify carboxypeptidase E (|CPE) and its splice variant, ΔN-CPE, as novel regulators of the Wnt pathway. We show that whereas ΔN-CPE activates the Wnt signal, the full-length CPE (F-CPE) protein is an inhibitor of Wnt/β-catenin signaling. F-CPE forms a complex with the Wnt3a ligand and the Frizzled receptor. Moreover, F-CPE disrupts disheveled-induced signalosomes that are important for transducing the Wnt signal and reduces β-catenin protein levels and activity. Taken together, our data indicate that F-CPE and ΔN-CPE regulate the canonical Wnt signaling pathway negatively and positively, respectively, and demonstrate that this screening approach can be a rapid means for isolation of novel Wnt signaling components.


Analytica Chimica Acta | 2010

Development of an immunoassay and a sol-gel-based immunoaffinity cleanup method for coplanar polychlorinated biphenyls from soil and sediment samples

Miriam Altstein; Orna Ben Aziz; Nir Skalka; Alisa Bronshtein; Jane C. Chuang; Jeanette M. Van Emon

Two polychlorinated biphenyls (PCB) enzyme linked immunosorbent assays (ELISAs) were developed using goat PCB purified immunoglobulin (IgG) antibodies (Abs). The IgGs exhibited the highest affinity toward PCB-77 (24 ng mL(-1)) with sensitivities in the range of 6-11 ng mL(-1). The Abs cross-reacted with PCB-126 and the heptachlorodibenzofuran 1,2,3,4,6,7,8-HpCDF but not with PCB-169, PCB-118, Aroclor 1232, 1248, 1260 or the hexachlorodibenzofuran 2,3,4,6,7,8-HxCDF. The IgGs were also used to develop a sol-gel-based immunoaffinity purification (IAP) method for cleanup of PCB-126. Recovery efficiencies depended on the sol-gel formats; a 1:12 format resulted in the highest binding capacity. Net binding capacity ranged from 112 to 257 ng, and 90% of the analyte could be eluted with only 2 mL of ethanol. The method was also very efficient in purifying PCB-126 from spiked soil and sediment samples from contaminated sites; and eliminating matrix interferences to a degree that enabled analysis of the purified samples by ELISA. The approaches developed in the course of the study form a basis for the development of additional IAP methods for other PCBs, and their implementation in high-throughput screening programs for PCB in food, soil, and other environmental and biological samples.


Molecular Cancer | 2014

Aldolase positively regulates of the canonical Wnt signaling pathway.

Michal Caspi; Gili Perry; Nir Skalka; Shilhav Meisel; Anastasia Firsow; Maayan Amit; Rina Rosin-Arbesfeld

The Wnt signaling pathway is an evolutionary conserved system, having pivotal roles during animal development. When over-activated, this signaling pathway is involved in cancer initiation and progression. The canonical Wnt pathway regulates the stability of β-catenin primarily by a destruction complex containing a number of different proteins, including Glycogen synthase kinase 3β (GSK-3β) and Axin, that promote proteasomal degradation of β-catenin. As this signaling cascade is modified by various proteins, novel screens aimed at identifying new Wnt signaling regulators were conducted in our laboratory. One of the different genes that were identified as Wnt signaling activators was Aldolase C (ALDOC). Here we report that ALDOC, Aldolase A (ALDOA) and Aldolase B (ALDOB) activate Wnt signaling in a GSK-3β-dependent mechanism, by disrupting the GSK-3β-Axin interaction and targeting Axin to the dishevelled (Dvl)-induced signalosomes that positively regulate the Wnt pathway thus placing the Aldolase proteins as novel Wnt signaling regulators.


Journal of Pharmaceutical and Biomedical Analysis | 2013

Development of generic immunoassay for the detection of a series of aminoglycosides with 6'-OH group for the treatment of genetic diseases in biological samples.

Moran Shalev; Jeyakumar Kandasamy; Nir Skalka; Valery Belakhov; Rina Rosin-Arbesfeld; Timor Baasov

Over the last two decades, a growing number of scientific evidences highlighted the potential therapeutic value of several structures of aminoglycoside antibiotics (including gentamicin and G418) for the treatment of various genetic diseases caused by nonsense mutations. These findings resulted in a fast evolvement of synthetic derivatives of aminoglycosides which were shown to be more target specific and less toxic than the clinically used antibiotics. The emerging progress in drug design and development has necessitated the urge to develop a fast, easy and accurate procedure for the determination of these potential therapeutic agents in various biologically derived matrices. Here we describe the preparation of a generic polyclonal antibody that was used for the development of homologous and heterologous immunoassays for the detection of a wide range of natural and synthetic aminoglycoside derivatives, highlighted today as potential therapeutic agents for the treatment of various genetic diseases. A common two-ring scaffold, NB82, present in the majority of compounds exhibiting potent biological activity, was used as a generic immunization hapten for the immunization of two rabbits. By using a series of chemical steps, NB82 was selectively conjugated via the N-1 position through glutaric acid linker to a carrier protein. Sensitivity (I₅₀) values for the recognition of three representative compounds NB82, NB84 and NB124 were determined to be 10 ± 3 ng mL⁻¹, 0.5 ± 0.04 μg mL⁻¹ and 1 ± 0.12 μg mL⁻¹, respectively. Limits of detection were determined to be 1 ± 0.3 ng mL⁻¹ for NB82, 20 ± 7 ng mL⁻¹ for NB84 and 15 ± 8 ng mL⁻¹ for NB124. The developed assays were further exploited for the in vivo monitoring of the therapeutic compounds in mice serum. Serum experimentations exhibited similar detection limits as observed for the PBS calibration experiments, demonstrating no interference with assays sensitivity, with rather high recovery ratios ranging from 92 to 107% in whole blood samples.


Molecular and Cellular Neuroscience | 2015

Neurotrophic factor-α1 modulates NGF-induced neurite outgrowth through interaction with Wnt-3a and Wnt-5a in PC12 cells and cortical neurons.

Prabhuanand Selvaraj; Jane S.W. Huang; Alexander Chen; Nir Skalka; Rina Rosin-Arbesfeld; Y. Peng Loh

Wnt-3a and Wnt-5a signaling activities inhibit and promote neurite outgrowth, respectively, to regulate dendritic and axonal genesis during neurodevelopment. NF-α1, a neurotrophic factor, has been shown to modulate dendritic remodeling and negatively regulate the canonical Wnt-3a pathway. Here, we investigated whether NF-α1 could modify nerve growth factor (NGF)-induced neurite outgrowth through interaction with Wnt-3a and Wnt-5a in PC12 cells and mouse primary cortical neurons. We showed that NGF-induced neurite outgrowth was inhibited by Wnt-3a, and this inhibition was prevented by NF-α1. Western blot analysis revealed that NF-α1 reduced the expression of both β-catenin in the canonical Wnt-3a pathway and Rho, a downstream effector of Wnt-3as non-canonical signaling pathway. Treatment of PC12 cells with a ROCK inhibitor prevented the inhibition of NGF-induced neurite outgrowth by Wnt-3a, suggesting that NF-α1 promotes neurite outgrowth in the presence of Wnt-3a by down-regulating its canonical and non-canonical activities. Interestingly, treatment of PC12 cells with Wnt-5a, which formed a complex with NF-α1, induced neurite outgrowth that was enhanced by treatment with the combination of Wnt-5a, NGF, and NF-α1. These effects of NF-α1 on Wnt 3as and Wnt 5as regulation of neurite outgrowth in PC12 cells were also demonstrated in primary cultures of mouse cortical neurons. In addition, we showed in PC12 cells that NF-α1 acts by upregulating adenomatous polyposis coli (APC) accumulation at neurite tips, thereby providing positive and negative Wnt-3a/Wnt-5a mediated cues to modulate neurite outgrowth, a process important during neurodevelopment.


Journal of Molecular Medicine | 2016

Erratum to: A flow cytometry-based reporter assay identifies macrolide antibiotics as nonsense mutation read-through agents

Michal Caspi; Anastasia Firsow; Raja Rajkumar; Nir Skalka; Itay Moshkovits; Ariel Munitz; Metsada Pasmanik-Chor; Hagar Greif; Dalia Megiddo; Revital Kariv; Daniel W. Rosenberg; Rina Rosin-Arbesfeld

A large number of human diseases are caused by nonsense mutations. These mutations result in premature protein termination and the expression of truncated, usually nonfunctional products. A promising therapeutic strategy for patients suffering from premature termination codon (PTC)-mediated disorders is to suppress the nonsense mutation and restore the expression of the affected protein. Such a suppression approach using specific antibiotics and other read-through promoting agents has been shown to suppress PTCs and restore the production of several important proteins. Here, we report the establishment of a novel, rapid, and very efficient method for screening stop-codon read-through agents. We also show that, in both mammalian cells and in a transgenic mouse model, distinct members of the macrolide antibiotic family can induce read-through of disease-causing stop codons leading to reexpression of several key proteins and to reduced disease phenotypes. Taken together, our results may help in the identification and characterization of well-needed customized pharmaceutical PTC suppression agents. Key messages & Establishment of a flow cytometry-based reporter assay to identify nonsense mutation read-through agents. & Macrolide antibiotics can induce read-through of diseasecausing stop codons. & Macrolide-induced protein restoration can alleviate disease-like phenotypes.


Oncogene | 2018

Klotho suppresses colorectal cancer through modulation of the unfolded protein response

Tammi Rubinstein; Shiri Shahmoon; Ehud Zigmond; Tal Etan; Keren Merenbakh-Lamin; Metsada Pasmanik-Chor; Gil Har-Zahav; Iris Barshack; Gilad W. Vainer; Nir Skalka; Rina Rosin-Arbesfeld; Chen Varol; Tami Rubinek; Ido Wolf

Klotho is an anti-aging transmembrane protein, which can be shed and function as a hormone. Accumulating data indicate klotho as a tumor suppressor in a wide array of malignancies and indicate the subdomain KL1 as the active region of the protein. We aimed to study the role of klotho as a tumor suppressor in colorectal cancer. Bioinformatics analyses of TCGA datasets indicated reduced klotho mRNA levels in human colorectal cancer, along with negative regulation of klotho expression by hypermethylation of the promoter and 1st exon, and hypomethylation of an area within the gene. Overexpression or treatment with klotho or KL1 inhibited proliferation of colorectal cancer cells in vitro. The in vivo activity of klotho and KL1 was examined using two models recapitulating development of tumors in the normal colonic environment of immune-competent mice. Treatment with klotho inhibited formation of colon polyps induced by the carcinogen azoxymethane, and KL1 treatment slowed growth of orthotopically-implanted colorectal tumors. Gene expression array revealed that klotho and KL1 expression enhanced the unfolded protein response (UPR) and this was further established by increased levels of spliced XBP1, GRP78 and phosphorylated-eIF2α. Furthermore, attenuation of the UPR partially abrogated klotho tumor suppressor activity. In conclusion, this study indicates klotho as a tumor suppressor in colorectal cancer and identifies, for the first time, the UPR as a pathway mediating klotho activities in cancer. These data suggest that administration of exogenous klotho or KL1 may serve as a novel strategy for prevention and treatment of colorectal cancer.


Cancer Research | 2015

Abstract 4959: Klotho suppresses colon cancer through modulation of the Wnt pathway and unfolded protein response

Tammi Rubinstein; Shiri Shahmoon; Gil Har Zahav; Nir Skalka; Tal Etan; Rina Arbesfeld; Metsada Pasmanik-Chor; Tami Rubinek; Ido Wolf

Aberrant activation of the canonical Wnt signal transduction pathway is implicated in the development and progression of colorectal cancer (CC). The key effector protein of this pathway is β-catenin, which functions with T-cell factor/lymphoid enhancer factor (TCF/LEF) to activate expression of Wnt target genes. Klotho is a transmembrane protein, which can be cleaved, shed and act as a circulating hormone. Klotho-deficient mice manifest a syndrome resembling accelerated aging, while klotho overexpression extends life span. We have previously identified klotho as a potent tumor suppressor in breast and pancreatic cancer, and recent data indicate it as a potent tumor suppressor in a wide array of malignancies, including CC. The activities of klotho include inhibition of the insulin like growth factor (IGF)-1 and the Wnt pathways. We aimed to decipher the role of klotho as a tumor suppressor and modulator of the Wnt pathway in CC. We first tested the effect of klotho on proliferation of CC cells and noted that overexpression of klotho inhibited colony formation of the HCT 116, SW480, HT-29, Colo-320 and RKO CC cells. We next examined the effects of klotho in vivo, using the azoxymethane (AOM) model, and found that daily administration of klotho to mice inhibited formation of colon polyps induced by AOM. While we did not see an effect of klotho on the IGF-1 signaling pathway, klotho did affect the Wnt pathway. Klotho reduced Wnt3A and β-catenin protein levels as measured by Western blotting, and inhibited transcriptional activity of the Wnt pathway as evidenced in luciferase assay. Moreover, this transcriptional inhibition was abrogated by overexpression of a constitutively active β-catenin. We, therefore, suspected that klotho inhibited the pathway upstream of β-catenin. Indeed, co-immunoprecipitation analyses indicated direct interaction between klotho and Wnt3A. As the inhibitory effect of klotho on colony formation of CC cells was only partially rescued by transfection with constitutively active β-catenin, we concluded that additional mechanisms may mediate the effects of klotho. In order to elucidate these mechanisms, we conducted a cDNA expression microarray and involvement of klotho in endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) was observed. Taken together, our data indicate klotho as a potent tumor suppressor in CC and suggest its role already at the early stages of polyp formation. Klotho effects in CC are partially mediated by the Wnt pathway however additional mechanisms, including regulation of ER stress response, are involved. Citation Format: Tammi Rubinstein, Shiri Shahmoon, Gil Har Zahav, Nir Skalka, Tal Etan, Rina Arbesfeld, Metsada Pasmanik-Chor, Tami Rubinek, Ido Wolf. Klotho suppresses colon cancer through modulation of the Wnt pathway and unfolded protein response. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 4959. doi:10.1158/1538-7445.AM2015-4959


Analytical and Bioanalytical Chemistry | 2011

Monitoring of the non-steroid anti-inflammatory drug indomethacin: development of immunochemical methods for its purification and detection

Nir Skalka; Alex Krol; Haim Schlesinger; Miriam Altstein

Collaboration


Dive into the Nir Skalka's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ido Wolf

Tel Aviv Sourasky Medical Center

View shared research outputs
Top Co-Authors

Avatar

Miriam Altstein

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Shiri Shahmoon

Tel Aviv Sourasky Medical Center

View shared research outputs
Top Co-Authors

Avatar

Tal Etan

Tel Aviv Sourasky Medical Center

View shared research outputs
Top Co-Authors

Avatar

Tami Rubinek

Tel Aviv Sourasky Medical Center

View shared research outputs
Top Co-Authors

Avatar

Tammi Rubinstein

Tel Aviv Sourasky Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge